Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
Proc Natl Acad Sci U S A ; 111(11): 4013-8, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591589

ABSTRACT

Alzheimer's disease (AD) is a common, progressive neurodegenerative disorder without effective disease-modifying therapies. The accumulation of amyloid-ß peptide (Aß) is associated with AD. However, identifying new compounds that antagonize the underlying cellular pathologies caused by Aß has been hindered by a lack of cellular models amenable to high-throughput chemical screening. To address this gap, we use a robust and scalable yeast model of Aß toxicity where the Aß peptide transits through the secretory and endocytic compartments as it does in neurons. The pathogenic Aß 1-42 peptide forms more oligomers and is more toxic than Aß 1-40 and genome-wide genetic screens identified genes that are known risk factors for AD. Here, we report an unbiased screen of ∼140,000 compounds for rescue of Aß toxicity. Of ∼30 hits, several were 8-hydroxyquinolines (8-OHQs). Clioquinol (CQ), an 8-OHQ previously reported to reduce Aß burden, restore metal homeostasis, and improve cognition in mouse AD models, was also effective and rescued the toxicity of Aß secreted from glutamatergic neurons in Caenorhabditis elegans. In yeast, CQ dramatically reduced Aß peptide levels in a copper-dependent manner by increasing degradation, ultimately restoring endocytic function. This mirrored its effects on copper-dependent oligomer formation in vitro, which was also reversed by CQ. This unbiased screen indicates that copper-dependent Aß oligomer formation contributes to Aß toxicity within the secretory/endosomal pathways where it can be targeted with selective metal binding compounds. Establishing the ability of the Aß yeast model to identify disease-relevant compounds supports its further exploitation as a validated early discovery platform.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Clioquinol/pharmacology , Endocytosis/physiology , Proteolysis/drug effects , Amyloid beta-Peptides/toxicity , Animals , Caenorhabditis elegans , Drug Discovery/methods , Endocytosis/drug effects , Small Molecule Libraries , Yeasts
3.
Mol Cell ; 30(6): 732-42, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18570876

ABSTRACT

The mechanisms of pre-mRNA splicing regulation are poorly understood. Here we dissect how the Saccharomyces cerevisiae ribosomal L30 protein blocks splicing of its pre-mRNA upon binding a kink-turn structure including the 5' splice site. We show that L30 binds the nascent RPL30 transcript without preventing recognition of the 5' splice site by U1 snRNP but blocking U2 snRNP association with the branch site. Interaction of the factors BBP and Mud2 with the intron, relevant for U2 snRNP recruitment, is not affected by L30. Furthermore, the functions of neither the DEAD-box protein Sub2 in the incipient spliceosome nor the U2 snRNP factor Cus2 on branch site recognition are required for L30 inhibition. These findings contrast with the effects caused by binding a heterologous protein to the same region, completely blocking intron recognition. Collectively, our data suggest that L30 represses a spliceosomal rearrangement required for U2 snRNP association with the transcript.


Subject(s)
Galectin 3/pharmacology , RNA Splicing , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Base Sequence , Introns , Models, Molecular , Nucleic Acid Conformation , Protein Binding , RNA, Fungal/chemistry , RNA, Fungal/drug effects , RNA, Fungal/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
4.
Mov Disord ; 29(10): 1231-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25131316

ABSTRACT

No disease-modifying therapies are available for synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple systems atrophy (MSA). The lack of therapies has been impeded by a paucity of validated drug targets and problematic cell-based model systems. New approaches are therefore needed to identify genes and compounds that directly target the underlying cellular pathologies elicited by the pathological protein, α-synuclein (α-syn). This small, lipid-binding protein impinges on evolutionarily conserved processes such as vesicle trafficking and mitochondrial function. For decades, the genetically tractable, single-cell eukaryote, budding yeast, has been used to study nearly all aspects of cell biology. More recently, yeast has revealed key insights into the underlying cellular pathologies caused by α-syn. The robust cellular toxicity caused by α-syn expression facilitates unbiased high-throughput small-molecule screening. Critically, one must validate the discoveries made in yeast in disease-relevant neuronal models. Here, we describe two recent reports that together establish yeast-to-human discovery platforms for synucleinopathies. In this exemplar, genes and small molecules identified in yeast were validated in patient-derived neurons that present the same cellular phenotypes initially discovered in yeast. On validation, we returned to yeast, where unparalleled genetic approaches facilitated the elucidation of a small molecule's mode of action. This approach enabled the identification and neuronal validation of a previously unknown "druggable" node that interfaces with the underlying, precipitating pathologies caused by α-syn. Such platforms can provide sorely needed leads and fresh ideas for disease-modifying therapy for these devastating diseases.


Subject(s)
Movement Disorders/pathology , Neurons/metabolism , Translational Research, Biomedical , Yeasts , alpha-Synuclein/metabolism , Animals , Humans , Movement Disorders/genetics , Yeasts/genetics , Yeasts/metabolism , alpha-Synuclein/genetics
5.
PLoS Biol ; 9(4): e1001052, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21541368

ABSTRACT

FUS/TLS is a nucleic acid binding protein that, when mutated, can cause a subset of familial amyotrophic lateral sclerosis (fALS). Although FUS/TLS is normally located predominantly in the nucleus, the pathogenic mutant forms of FUS/TLS traffic to, and form inclusions in, the cytoplasm of affected spinal motor neurons or glia. Here we report a yeast model of human FUS/TLS expression that recapitulates multiple salient features of the pathology of the disease-causing mutant proteins, including nuclear to cytoplasmic translocation, inclusion formation, and cytotoxicity. Protein domain analysis indicates that the carboxyl-terminus of FUS/TLS, where most of the ALS-associated mutations are clustered, is required but not sufficient for the toxicity of the protein. A genome-wide genetic screen using a yeast over-expression library identified five yeast DNA/RNA binding proteins, encoded by the yeast genes ECM32, NAM8, SBP1, SKO1, and VHR1, that rescue the toxicity of human FUS/TLS without changing its expression level, cytoplasmic translocation, or inclusion formation. Furthermore, hUPF1, a human homologue of ECM32, also rescues the toxicity of FUS/TLS in this model, validating the yeast model and implicating a possible insufficiency in RNA processing or the RNA quality control machinery in the mechanism of FUS/TLS mediated toxicity. Examination of the effect of FUS/TLS expression on the decay of selected mRNAs in yeast indicates that the nonsense-mediated decay pathway is probably not the major determinant of either toxicity or suppression.


Subject(s)
DNA Helicases/metabolism , RNA-Binding Protein FUS/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Trans-Activators/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cell Nucleus/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , DNA Helicases/genetics , Gene Expression Regulation , Mutation , Neurons/metabolism , RNA Helicases , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein FUS/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
6.
J Biol Chem ; 287(6): 4107-20, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22147697

ABSTRACT

No current therapies target the underlying cellular pathologies of age-related neurodegenerative diseases. Model organisms provide a platform for discovering compounds that protect against the toxic, misfolded proteins that initiate these diseases. One such protein, TDP-43, is implicated in multiple neurodegenerative diseases, including amyotrophic lateral sclerosis and frontotemporal lobar degeneration. In yeast, TDP-43 expression is toxic, and genetic modifiers first discovered in yeast have proven to modulate TDP-43 toxicity in both neurons and humans. Here, we describe a phenotypic screen for small molecules that reverse TDP-43 toxicity in yeast. One group of hit compounds was 8-hydroxyquinolines (8-OHQ), a class of clinically relevant bioactive metal chelators related to clioquinol. Surprisingly, in otherwise wild-type yeast cells, different 8-OHQs had selectivity for rescuing the distinct toxicities caused by the expression of TDP-43, α-synuclein, or polyglutamine proteins. In fact, each 8-OHQ synergized with the other, clearly establishing that they function in different ways. Comparative growth and molecular analyses also revealed that 8-OHQs have distinct metal chelation and ionophore activities. The diverse bioactivity of 8-OHQs indicates that altering different aspects of metal homeostasis and/or metalloprotein activity elicits distinct protective mechanisms against several neurotoxic proteins. Indeed, phase II clinical trials of an 8-OHQ has produced encouraging results in modifying Alzheimer disease. Our unbiased identification of 8-OHQs in a yeast TDP-43 toxicity model suggests that tailoring 8-OHQ activity to a particular neurodegenerative disease may be a viable therapeutic strategy.


Subject(s)
DNA-Binding Proteins/metabolism , Models, Biological , Oxyquinoline/pharmacology , Polyglutamic Acid/metabolism , TDP-43 Proteinopathies/drug therapy , alpha-Synuclein/metabolism , Animals , Caenorhabditis elegans , Chelating Agents/pharmacology , DNA-Binding Proteins/genetics , Humans , Ionophores/pharmacology , Polyglutamic Acid/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , alpha-Synuclein/genetics
7.
Drug Discov Today Technol ; 10(1): e121-8, 2013.
Article in English | MEDLINE | ID: mdl-24050240

ABSTRACT

Parkinson's disease (PD) is a devastating neurodegenerative disease that affects over one million patients in the US. Yet, no disease modifying drugs exist, only those that temporarily alleviate symptoms. Because of its poorly defined and highly complex disease etiology, it is essential to embrace unbiased and innovative approaches for identifying new chemical entities that target the underlying toxicities associated with PD. Traditional target-based drug discovery paradigm can suffer from a bias toward a small number of potential targets. Phenotypic screening of both genetic and pharmacological PD models offers an alternative approach to discover compounds that target the initiating causes and effectors of cellular toxicity. The relative paucity of reported phenotypic screens illustrates the intrinsic difficulty in establishing model systems that are both biologically meaningful and adaptable to high-throughput screening. Parallel advances in PD models and in vivo screening technologies will help create opportunities for identifying new therapeutic leads with unanticipated, breakthrough mechanisms of action.


Subject(s)
Drug Evaluation, Preclinical , Parkinson Disease/drug therapy , Animals , High-Throughput Screening Assays , Humans , Phenotype , Yeasts/genetics , alpha-Synuclein/genetics
8.
Sci Transl Med ; 15(679): eabq6288, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36652537

ABSTRACT

Deregulated de novo lipid synthesis (DNLS) is a potential druggable vulnerability in glioblastoma (GBM), a highly lethal and incurable cancer. Yet the molecular mechanisms that determine susceptibility to DNLS-targeted therapies remain unknown, and the lack of brain-penetrant inhibitors of DNLS has prevented their clinical evaluation as GBM therapeutics. Here, we report that YTX-7739, a clinical-stage inhibitor of stearoyl CoA desaturase (SCD), triggers lipotoxicity in patient-derived GBM stem-like cells (GSCs) and inhibits fatty acid desaturation in GSCs orthotopically implanted in mice. When administered as a single agent, or in combination with temozolomide (TMZ), YTX-7739 showed therapeutic efficacy in orthotopic GSC mouse models owing to its lipotoxicity and ability to impair DNA damage repair. Leveraging genetic, pharmacological, and physiological manipulation of key signaling nodes in gliomagenesis complemented with shotgun lipidomics, we show that aberrant MEK/ERK signaling and its repression of the energy sensor AMP-activated protein kinase (AMPK) primarily drive therapeutic vulnerability to SCD and other DNLS inhibitors. Conversely, AMPK activation mitigates lipotoxicity and renders GSCs resistant to the loss of DNLS, both in culture and in vivo, by decreasing the saturation state of phospholipids and diverting toxic lipids into lipid droplets. Together, our findings reveal mechanisms of metabolic plasticity in GSCs and provide a framework for the rational integration of DNLS-targeted GBM therapies.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Glioblastoma/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/therapeutic use , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Xenograft Model Antitumor Assays , DNA Damage , Lipids , Neoplastic Stem Cells/metabolism
9.
Mol Neurobiol ; 59(4): 2171-2189, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35060064

ABSTRACT

Stearoyl-CoA desaturase (SCD) is a potential therapeutic target for Parkinson's and related neurodegenerative diseases. SCD inhibition ameliorates neuronal toxicity caused by aberrant α-synuclein, a lipid-binding protein implicated in Parkinson's disease. Its inhibition depletes monounsaturated fatty acids, which may modulate α-synuclein conformations and membrane interactions. Herein, we characterize the pharmacokinetic and pharmacodynamic properties of YTX-7739, a clinical-stage SCD inhibitor. Administration of YTX-7739 to rats and monkeys for 15 days caused a dose-dependent increase in YTX-7739 concentrations that were well-tolerated and associated with concentration-dependent reductions in the fatty acid desaturation index (FADI), the ratio of monounsaturated to saturated fatty acids. An approximate 50% maximal reduction in the carbon-16 desaturation index was observed in the brain, with comparable responses in the plasma and skin. A study with a diet supplemented in SCD products indicates that changes in brain C16 desaturation were due to local SCD inhibition, rather than to changes in systemic fatty acids that reach the brain. Assessment of pharmacodynamic response onset and reversibility kinetics indicated that approximately 7 days of dosing were required to achieve maximal responses, which persisted for at least 2 days after cessation of dosing. YTX-7739 thus achieved sufficient concentrations in the brain to inhibit SCD and produce pharmacodynamic responses that were well-tolerated in rats and monkeys. These results provide a framework for evaluating YTX-7739 pharmacology clinically as a disease-modifying therapy to treat synucleinopathies.


Subject(s)
Parkinson Disease , Stearoyl-CoA Desaturase , Animals , Fatty Acids/metabolism , Fatty Acids/pharmacology , Lipid Metabolism/physiology , Rats , Stearoyl-CoA Desaturase/metabolism , alpha-Synuclein/metabolism
10.
Neurotherapeutics ; 19(3): 1018-1036, 2022 04.
Article in English | MEDLINE | ID: mdl-35445353

ABSTRACT

Increasing evidence has shown that Parkinson's disease (PD) impairs midbrain dopaminergic, cortical and other neuronal subtypes in large part due to the build-up of lipid- and vesicle-rich α-synuclein (αSyn) cytotoxic inclusions. We previously identified stearoyl-CoA desaturase (SCD) as a potential therapeutic target for synucleinopathies. A brain-penetrant SCD inhibitor, YTX-7739, was developed and has entered Phase 1 clinical trials. Here, we report the efficacy of YTX-7739 in reversing pathological αSyn phenotypes in various in vitro and in vivo PD models. In cell-based assays, YTX-7739 decreased αSyn-mediated neuronal death, reversed the abnormal membrane interaction of amplified E46K ("3K") αSyn, and prevented pathological phenotypes in A53T and αSyn triplication patient-derived neurospheres, including dysregulated fatty acid profiles and pS129 αSyn accumulation. In 3K PD-like mice, YTX-7739 crossed the blood-brain barrier, decreased unsaturated fatty acids, and prevented progressive motor deficits. Both YTX-7739 treatment and decreasing SCD activity through deletion of one copy of the SCD1 gene (SKO) restored the physiological αSyn tetramer-to-monomer ratio, dopaminergic integrity, and neuronal survival in 3K αSyn mice. YTX-7739 efficiently reduced pS129 + and PK-resistant αSyn in both human wild-type αSyn and 3K mutant mice similar to the level of 3K-SKO. Together, these data provide further validation of SCD as a PD therapeutic target and YTX-7739 as a clinical candidate for treating human α-synucleinopathies.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Brain/metabolism , Humans , Mice , Neurons/metabolism , Parkinson Disease/genetics , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
11.
Proc Natl Acad Sci U S A ; 104(50): 19948-53, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077427

ABSTRACT

To characterize proteins associated with active transcription complexes, we purified RNA polymerase II (pol II) from Saccharomyces cerevisiae after fixing live cells with formaldehyde. The approach mimics ChIP and requires solubilizing cross-linked complexes with sonication. Pol II was affinity-purified, and associated proteins were identified by MS. Several classes of proteins depended on cross-linking, including Mediator, general transcription factors, elongation factors, ribonucleoprotein particle (RNP) proteins, and histones. A tagged RNP protein reciprocally purified pol II under identical cross-linking conditions, and the association between RNP proteins and pol II was largely RNase-sensitive. The data indicate that the cross-linked Pol II purification contains elongating pol II with associated nascent RNP. Consistent with this view, some elongation factors no longer associate with pol II after inactivation of transcription in the temperature-sensitive pol II mutant, rpb1-1. Taken together, our data suggest that the cross-linked pol II purification contains a mixed population of pol II, including initiating pol II and elongating pol II.


Subject(s)
Fungal Proteins/chemistry , Protein Subunits/chemistry , RNA Polymerase II/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Cross-Linking Reagents , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Oligonucleotide Array Sequence Analysis/methods , Protein Subunits/genetics , Protein Subunits/isolation & purification , RNA Polymerase II/genetics , RNA Polymerase II/isolation & purification , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , Ribonucleoproteins , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Tandem Mass Spectrometry , Transcriptional Activation/genetics
12.
Methods Mol Biol ; 2049: 419-444, 2019.
Article in English | MEDLINE | ID: mdl-31602625

ABSTRACT

Neurodegenerative diseases (ND) represent a growing, global health crisis, one that lacks any disease-modifying therapeutic strategy. This critical need for new therapies must be met with an exhaustive approach to exploit all tools available. A yeast (Saccharomyces cerevisiae) model of α-synuclein toxicity-the protein causally linked to Parkinson's disease and other synucleinopathies-offers a powerful approach that takes advantage of the unique offerings of this system: tractable genetics, robust high-throughput screening strategies, unparalleled data repositories, powerful computational tools, and extensive evolutionary conservation of fundamental biological pathways. These attributes have enabled genetic and small molecule screens that have revealed toxic phenotypes and drug targets that translate directly to patient-derived iPSC neurons. Extending these insights, recent advances in genetic network analyses have generated the first "humanized" α-synuclein network, which has identified druggable proteins and led to validation of the toxic phenotypes in patient-derived cells. Unbiased phenotypic small molecule screens can identify compounds targeting critical proteins within α-synuclein networks. While identification of direct drug targets for phenotypic screen hits represents a bottleneck, high-throughput chemical genetic methods provide a means to uncover cellular targets and pathways for large numbers of compounds in parallel. Taken together, the yeast α-synuclein model and associated tools can reveal insights into underlying cellular pathologies, lead molecules and their cognate targets, and strategies to translate mechanisms of toxicity and cytoprotection into complex neuronal systems.


Subject(s)
Saccharomyces cerevisiae/metabolism , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , Animals , Drug Evaluation, Preclinical , Gene Regulatory Networks , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Synucleinopathies/drug therapy
13.
Cell Rep ; 25(10): 2742-2754.e31, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517862

ABSTRACT

The lack of disease-modifying treatments for neurodegenerative disease stems in part from our rudimentary understanding of disease mechanisms and the paucity of targets for therapeutic intervention. Here we used an integrated discovery paradigm to identify a new therapeutic target for diseases caused by α-synuclein (α-syn), a small lipid-binding protein that misfolds and aggregates in Parkinson's disease and other disorders. Using unbiased phenotypic screening, we identified a series of compounds that were cytoprotective against α-syn-mediated toxicity by inhibiting the highly conserved enzyme stearoyl-CoA desaturase (SCD). Critically, reducing the levels of unsaturated membrane lipids by inhibiting SCD reduced α-syn toxicity in human induced pluripotent stem cell (iPSC) neuronal models. Taken together, these findings suggest that inhibition of fatty acid desaturation has potential as a therapeutic approach for the treatment of Parkinson's disease and other synucleinopathies.


Subject(s)
Stearoyl-CoA Desaturase/antagonists & inhibitors , alpha-Synuclein/toxicity , Animals , Cytoprotection/drug effects , Fatty Acids/metabolism , Humans , Lipid Metabolism/drug effects , Neurons/drug effects , Neurons/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Protein Aggregates , Rats , Saccharomyces cerevisiae/drug effects , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism
14.
ACS Chem Neurosci ; 8(9): 2039-2055, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28628299

ABSTRACT

The lack of therapies for neurodegenerative diseases arises from our incomplete understanding of their underlying cellular toxicities and the limited number of predictive model systems. It is critical that we develop approaches to identify novel targets and lead compounds. Here, a phenotypic screen of yeast proteinopathy models identified dihydropyrimidine-thiones (DHPM-thiones) that selectively rescued the toxicity caused by ß-amyloid (Aß), the peptide implicated in Alzheimer's disease. Rescue of Aß toxicity by DHPM-thiones occurred through a metal-dependent mechanism of action. The bioactivity was distinct, however, from that of the 8-hydroxyquinoline clioquinol (CQ). These structurally dissimilar compounds strongly synergized at concentrations otherwise not competent to reduce toxicity. Cotreatment ameliorated Aß toxicity by reducing Aß levels and restoring functional vesicle trafficking. Notably, these low doses significantly reduced deleterious off-target effects caused by CQ on mitochondria at higher concentrations. Both single and combinatorial treatments also reduced death of neurons expressing Aß in a nematode, indicating that DHPM-thiones target a conserved protective mechanism. Furthermore, this conserved activity suggests that expression of the Aß peptide causes similar cellular pathologies from yeast to neurons. Our identification of a new cytoprotective scaffold that requires metal-binding underscores the critical role of metal phenomenology in mediating Aß toxicity. Additionally, our findings demonstrate the valuable potential of synergistic compounds to enhance on-target activities, while mitigating deleterious off-target effects. The identification and prosecution of synergistic compounds could prove useful for developing AD therapeutics where combination therapies may be required to antagonize diverse pathologies.


Subject(s)
Amyloid beta-Peptides/metabolism , Clioquinol/pharmacology , Metals/metabolism , Neuroprotective Agents/pharmacology , Thiones/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Clioquinol/toxicity , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , Homeostasis/drug effects , Homeostasis/physiology , Ions/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuroprotective Agents/toxicity , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Thiones/toxicity , Yeasts
16.
Cell Syst ; 4(2): 157-170.e14, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28131822

ABSTRACT

Numerous genes and molecular pathways are implicated in neurodegenerative proteinopathies, but their inter-relationships are poorly understood. We systematically mapped molecular pathways underlying the toxicity of alpha-synuclein (α-syn), a protein central to Parkinson's disease. Genome-wide screens in yeast identified 332 genes that impact α-syn toxicity. To "humanize" this molecular network, we developed a computational method, TransposeNet. This integrates a Steiner prize-collecting approach with homology assignment through sequence, structure, and interaction topology. TransposeNet linked α-syn to multiple parkinsonism genes and druggable targets through perturbed protein trafficking and ER quality control as well as mRNA metabolism and translation. A calcium signaling hub linked these processes to perturbed mitochondrial quality control and function, metal ion transport, transcriptional regulation, and signal transduction. Parkinsonism gene interaction profiles spatially opposed in the network (ATP13A2/PARK9 and VPS35/PARK17) were highly distinct, and network relationships for specific genes (LRRK2/PARK8, ATXN2, and EIF4G1/PARK18) were confirmed in patient induced pluripotent stem cell (iPSC)-derived neurons. This cross-species platform connected diverse neurodegenerative genes to proteinopathy through specific mechanisms and may facilitate patient stratification for targeted therapy.


Subject(s)
Neurodegenerative Diseases/pathology , alpha-Synuclein/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Ataxin-2/chemistry , Ataxin-2/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Susceptibility , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Gene Regulatory Networks/genetics , Genome, Fungal , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurodegenerative Diseases/genetics , Neurons/cytology , Neurons/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , alpha-Synuclein/genetics
17.
Nucleic Acids Res ; 31(8): 2234-41, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682374

ABSTRACT

To investigate the stringency of the Escherichia coli selenocysteine insertion sequence (SECIS) requirements, libraries of SECIS variants were screened via a novel method in which suppression of the selenocysteine (Sec) opal codon was coupled to bacteriophage plaque formation. The SECIS variant libraries were designed with a mostly paired lower stem, so that randomization could be focused on the upper stem and loop regions. We identified 19 functional non-native SECIS sequences that violated the expected pairing requirements for the SECIS upper stem. All of the SECIS variants were shown to permit Sec insertion in phage (by chemical modification of the Sec residue) and fused to lacZalpha (by beta-galactosidase assay). The diminished pairing of the upper stem appears to be mitigated by the overall stem stability; a given upper stem variant has significantly higher readthrough in the context of a paired, rather than unpaired, lower stem. These results suggest an unexpected downstream sequence flexibility in prokaryotic selenoprotein expression.


Subject(s)
Escherichia coli/genetics , Gene Library , Regulatory Sequences, Nucleic Acid/genetics , Selenocysteine/genetics , Bacteriophages/genetics , Base Sequence , Cloning, Molecular , Codon, Terminator/genetics , DNA, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Lac Operon/genetics , Mutation , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Selenocysteine/metabolism , Sodium Selenite/pharmacology , beta-Galactosidase/drug effects , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
18.
Nat Rev Neurol ; 11(6): 339-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25986505

ABSTRACT

In the absence of a single preventive or disease-modifying strategy, neurodegenerative diseases are becoming increasingly prevalent in our ageing population. The mechanisms underlying neurodegeneration are poorly understood, making the target-based drug screening strategies that are employed by the pharmaceutical industry fraught with difficulty. However, phenotypic screening in neurons and glia derived from patients is now conceivable through unprecedented developments in reprogramming, transdifferentiation, and genome editing. We outline progress in this nascent field, but also consider the formidable hurdles to identifying robust, disease-relevant and screenable cellular phenotypes in patient-derived cells. We illustrate how analysis in the simple baker's yeast cell Saccharaomyces cerevisiae is driving discovery in patient-derived neurons, and how approaches in this model organism can establish a paradigm to guide the development of stem cell-based phenotypic screens.


Subject(s)
Genetic Testing/methods , Neurodegenerative Diseases/genetics , Genetic Predisposition to Disease , Humans , Models, Genetic , Models, Neurological , Neuroglia , Neurons , Saccharomyces cerevisiae/genetics , Stem Cells
19.
Biochem Mol Biol Educ ; 32(4): 239-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-21706731

ABSTRACT

We have developed an introductory bioinformatics exercise for sophomore biology and biochemistry students that reinforces the understanding of the structure of a gene and the principles and events involved in its expression. In addition, the activity illustrates the severe effect mutations in a gene sequence can have on the protein product. Students search GenBank for the wild-type nucleotide sequence of the Caenorhabditis elegans unc-22 gene, the amino acid sequence of its gene product, and the nucleotide sequence of the transposon Tc5. The nucleotide sequences are manipulated using two programs in the Lasergene® software package from DNASTAR®. The first program, EditSeq®, enables students to experience the meticulous process required to precisely locate and remove intron sequences from the wild-type unc-22 allele to generate a cDNA sequence. The unc-22(r466) allele is generated by inserting the sequence of the transposon Tc5 into the appropriate location of the third exon in unc-22. The open reading frames of both cDNAs are located and then translated. MegAlign®, the second program, aligns the wild-type sequence of the UNC-22 protein and the wild-type and mutant protein sequences that were constructed. The degree of sequence similarity between the aligned proteins allows students to verify their success in processing the gene, as well as to visualize the truncated protein product from the Tc5 mutant allele. Student feedback and possible modifications to the exercise as well as supplemental exercises are also discussed.

20.
Science ; 342(6161): 979-83, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24158909

ABSTRACT

α-Synuclein (α-syn) is a small lipid-binding protein implicated in several neurodegenerative diseases, including Parkinson's disease, whose pathobiology is conserved from yeast to man. There are no therapies targeting these underlying cellular pathologies, or indeed those of any major neurodegenerative disease. Using unbiased phenotypic screens as an alternative to target-based approaches, we discovered an N-aryl benzimidazole (NAB) that strongly and selectively protected diverse cell types from α-syn toxicity. Three chemical genetic screens in wild-type yeast cells established that NAB promoted endosomal transport events dependent on the E3 ubiquitin ligase Rsp5/Nedd4. These same steps were perturbed by α-syn itself. Thus, NAB identifies a druggable node in the biology of α-syn that can correct multiple aspects of its underlying pathology, including dysfunctional endosomal and endoplasmic reticulum-to-Golgi vesicle trafficking.


Subject(s)
Benzimidazoles/pharmacology , Cytoprotection , Endosomal Sorting Complexes Required for Transport/genetics , Gene Regulatory Networks/drug effects , Neurodegenerative Diseases/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/genetics , alpha-Synuclein/metabolism , Animals , Benzimidazoles/chemistry , Caenorhabditis elegans , Cells, Cultured , Drug Evaluation, Preclinical , Nedd4 Ubiquitin Protein Ligases , Neurons/metabolism , Parkinson Disease/metabolism , Rats , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL